1
|
Romeo I, Brizzi A, Pessina F, Ambrosio FA, Aiello F, Belardo C, Carullo G, Costa G, De Petrocellis L, Frosini M, Luongo L, Maramai S, Paolino M, Moriello AS, Mugnaini C, Scorzelli F, Maione S, Corelli F, Di Marzo V, Alcaro S, Artese A. In Silico-Guided Rational Drug Design and Synthesis of Novel 4-(Thiophen-2-yl)butanamides as Potent and Selective TRPV1 Agonists. J Med Chem 2023; 66:6994-7015. [PMID: 37192374 DOI: 10.1021/acs.jmedchem.3c00447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
We describe an in silico-guided rational drug design and the synthesis of the suggested ligands, aimed at improving the TRPV1-ligand binding properties and the potency of N-(4-hydroxy-3-methoxybenzyl)-4-(thiophen-2-yl) butanamide I, a previously identified TRPV1 agonist. The docking experiments followed by molecular dynamics simulations and thermodynamic analysis led the drug design toward both the introduction of a lipophilic iodine and a flat pyridine/benzene at position 5 of the thiophene nucleus. Most of the synthesized compounds showed high TRPV1 efficacy and potency as well as selectivity. The molecular modeling analysis highlighted crucial hydrophobic interactions between Leu547 and the iodo-thiophene nucleus, as in amide 2a, or between Phe543 and the pyridinyl moiety, as in 3a. In the biological evaluation, both compounds showed protective properties against oxidative stress-induced ROS formation in human keratinocytes. Additionally, while 2a showed neuroprotective effects in both neurons and rat brain slices, 3a exhibited potent antinociceptive effect in vivo..
Collapse
Affiliation(s)
- Isabella Romeo
- Dipartimento di Scienze della Salute, Università degli Studi "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, Università degli Studi "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
| | - Antonella Brizzi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Federica Pessina
- Dipartimento di Medicina Molecolare e dello Sviluppo, Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Francesca Alessandra Ambrosio
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
| | - Francesca Aiello
- Dipartimento di Farmacia e Scienza della Salute e della Nutrizione, Università della Calabria, Via P. Bucci, 87036 Arcavacata di Rende, Cosenza, Italy
| | - Carmela Belardo
- Dipartimento di Medicina Sperimentale, Divisione di Farmacologia, Università degli Studi della Campania "L. Vanvitelli", |Via Costantinopoli 16, 80138 Napoli, Italy
| | - Gabriele Carullo
- Dipartimento di Scienze della Vita, Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Giosuè Costa
- Dipartimento di Scienze della Salute, Università degli Studi "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, Università degli Studi "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
| | - Luciano De Petrocellis
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, Comprensorio Olivetti, 80078 Pozzuoli, Napoli, Italy
| | - Maria Frosini
- Dipartimento di Scienze della Vita, Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Livio Luongo
- Dipartimento di Medicina Sperimentale, Divisione di Farmacologia, Università degli Studi della Campania "L. Vanvitelli", |Via Costantinopoli 16, 80138 Napoli, Italy
| | - Samuele Maramai
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Marco Paolino
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Aniello Schiano Moriello
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, Comprensorio Olivetti, 80078 Pozzuoli, Napoli, Italy
- Epitech Group SpA, Via L. Einaudi 13, 35030 Saccolongo, Padova, Italy
| | - Claudia Mugnaini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Francesco Scorzelli
- Recipharm (Edmond Pharma), Strada Statale dei Giovi 131, 20037 Paderno Dugnano, Milano, Italy
| | - Sabatino Maione
- Dipartimento di Medicina Sperimentale, Divisione di Farmacologia, Università degli Studi della Campania "L. Vanvitelli", |Via Costantinopoli 16, 80138 Napoli, Italy
| | - Federico Corelli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, Comprensorio Olivetti, 80078 Pozzuoli, Napoli, Italy
- Heart and Lung Research Institute, Department of Medicine, Faculty of Medicine, and Institute of Nutrition and Functional Foods, NUTRISS Center, School of Nutrition, Faculty of Agriculture and Food Science, Université Laval, 2325 Rue de l'Université, Québec, Canada
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università degli Studi "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, Università degli Studi "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
| | - Anna Artese
- Dipartimento di Scienze della Salute, Università degli Studi "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
- Net4Science Academic Spin-Off, Università degli Studi "Magna Græcia" di Catanzaro, Campus "S. Venuta", Viale Europa, 88100 Catanzaro, Italy
| |
Collapse
|
2
|
Campbell-Yeo M, Benoit B, Newman A, Johnston C, Bardouille T, Stevens B, Jiang A. The influence of skin-to-skin contact on Cortical Activity during Painful procedures in preterm infants in the neonatal intensive care unit (iCAP mini): study protocol for a randomized control trial. Trials 2022; 23:512. [PMID: 35725632 PMCID: PMC9208173 DOI: 10.1186/s13063-022-06424-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/24/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Strong evidence suggests that maternal-infant skin-to-skin contact (SSC) is effective in reducing behavioural responses to pain. Given the multi-sensory benefits of SSC, it is highly likely that SSC provided during pain in early life may reduce pain-induced brain activity. The aim of this study is to examine the effect of SSC compared to 24% sucrose on pain-induced activity in the preterm infant brain during a medically required heel lance. Secondary objectives include determining (a) differences between behavioural pain response and noxious-related brain activity during heel lance and (b) rate of adverse events across groups. METHODS We will randomly assign 126 babies (32 to 36 completed weeks gestational age) admitted to the neonatal intensive care unit, and their mothers within the first seven days of age to receive (i) SSC plus sterile water and (ii) 24% oral sucrose. Each baby will receive a medically indicated heel lance, following a no treatment baseline period. The primary outcome is noxious-related brain activity measured using an electroencephalogram (EEG) pain-specific event-related potential. Secondary outcomes include pain intensity measured using a bio-behavioural infant pain assessment tool (Premature Infant Pain Profile-Revised) and rate of adverse events. DISCUSSION This will be the first clinical trial to compare the effect of SSC and 24% sucrose on pain-induced brain activity in the preterm infant brain during a clinical noxious stimulus, measured using EEG. Given the negative neurodevelopmental outcomes associated with unmanaged pain, it is imperative that preterm babies receive the most effective pain-reducing treatments to improve their health outcomes. Our findings will have important implications in informing optimal pain assessment and management in preterm infants. TRIAL REGISTRATION ClinicalTrials.gov NCT03745963 . Registered on November 19, 2018.
Collapse
Affiliation(s)
- Marsha Campbell-Yeo
- School of Nursing, Faculty of Health, Dalhousie University and IWK Health, Halifax, NS Canada
| | - Britney Benoit
- Rankin School of Nursing, St. Francis Xavier University, Antigonish, NS Canada
| | - Aaron Newman
- Faculty of Science, Department of Psychology & Neuroscience, Dalhousie University, Halifax, NS Canada
| | | | - Tim Bardouille
- Department of Physics & Atmospheric Science, Dalhousie University, Halifax, NS Canada
| | - Bonnie Stevens
- Lawrence S Bloomberg Faculty of Nursing, University of Toronto and Child Health Evaluative Sciences Program, Research Institute, The Hospital for Sick Children (SickKids), Toronto, ON Canada
| | | |
Collapse
|
3
|
Toussaint AB, Foster W, Jones JM, Kaufmann S, Wachira M, Hughes R, Bongiovanni AR, Famularo ST, Dunham BP, Schwark R, Karbalaei R, Dressler C, Bavley CC, Fried NT, Wimmer ME, Abdus-Saboor I. Chronic paternal morphine exposure increases sensitivity to morphine-derived pain relief in male progeny. SCIENCE ADVANCES 2022; 8:eabk2425. [PMID: 35171664 PMCID: PMC8849295 DOI: 10.1126/sciadv.abk2425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Parental history of opioid exposure is seldom considered when prescribing opioids for pain relief. To explore whether parental opioid exposure may affect sensitivity to morphine in offspring, we developed a "rat pain scale" with high-speed imaging, machine learning, and mathematical modeling in a multigenerational model of paternal morphine self-administration. We find that the most commonly used tool to measure mechanical sensitivity in rodents, the von Frey hair, is not painful in rats during baseline conditions. We also find that male progeny of morphine-treated sires had no baseline changes in mechanical pain sensitivity but were more sensitive to the pain-relieving effects of morphine. Using RNA sequencing across pain-relevant brain regions, we identify gene expression changes within the regulator of G protein signaling family of proteins that may underlie this multigenerational phenotype. Together, this rat pain scale revealed that paternal opioid exposure increases sensitivity to morphine's pain-relieving effects in male offspring.
Collapse
Affiliation(s)
- Andre B. Toussaint
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - William Foster
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Jessica M. Jones
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Samuel Kaufmann
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Meghan Wachira
- Department of Biology, Rutgers Camden University, Camden, NJ, USA
| | - Robert Hughes
- Department of Biology, Rutgers Camden University, Camden, NJ, USA
| | - Angela R. Bongiovanni
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Sydney T. Famularo
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Benjamin P. Dunham
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Ryan Schwark
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Reza Karbalaei
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Carmen Dressler
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Charlotte C. Bavley
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Nathan T. Fried
- Department of Biology, Rutgers Camden University, Camden, NJ, USA
| | - Mathieu E. Wimmer
- Department of Psychology, Program in Neuroscience Temple University, Philadelphia, PA, USA
| | - Ishmail Abdus-Saboor
- Zuckerman Mind Brain Behavior Institute and Department of Biological Sciences, Columbia University, New York, NY, USA
- Corresponding author.
| |
Collapse
|
4
|
Elliott JM, Rueckeis CA, Pan Y, Parrish TB, Walton DM, Linnstaedt SD. microRNA let-7i-5p mediates the relationship between muscle fat infiltration and neck pain disability following motor vehicle collision: a preliminary study. Sci Rep 2021; 11:3140. [PMID: 33542428 PMCID: PMC7862492 DOI: 10.1038/s41598-021-82734-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/11/2021] [Indexed: 01/30/2023] Open
Abstract
Persistent neck-pain disability (PNPD) is common following traumatic stress exposures such as motor vehicle collision (MVC). Substantial literature indicates that fat infiltration into neck muscle (MFI) is associated with post-MVC PNPD. However, little is known about the molecular mediators underlying this association. In the current study, we assessed whether microRNA expression signatures predict PNPD and whether microRNA mediate the relationship between neck MFI and PNPD. A nested cohort of 43 individuals from a longitudinal study of MVC survivors, who provided blood (PAXgene RNA) and underwent magnetic resonance imaging (MRI), were included in the current study. Peritraumatic microRNA expression levels were quantified via small RNA sequencing, neck MFI via MRI, and PNPD via the Neck Disability Index two-weeks, three-months, and twelve-months following MVC. Repeated measures regression models were used to assess the relationship between microRNA and PNPD and to perform mediation analyses. Seventeen microRNA predicted PNPD following MVC. One microRNA, let-7i-5p, mediated the relationship between neck MFI and PNPD. Peritraumatic blood-based microRNA expression levels predict PNPD following MVC and let-7i-5p might contribute to the underlying effects of neck MFI on persistent disability. In conclusion, additional studies are needed to validate this finding.
Collapse
Affiliation(s)
- James M Elliott
- Faculty of Medicine and Health, The Northern Sydney Local Health District, The Kolling Institute, The University of Sydney, St. Leonards, NSW, Australia
- Physical Therapy and Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Cathleen A Rueckeis
- Institute for Trauma Recovery, University of North Carolina, Campus Box #7010, Chapel Hill, NC, 27599-7010, USA
| | - Yue Pan
- Institute for Trauma Recovery, University of North Carolina, Campus Box #7010, Chapel Hill, NC, 27599-7010, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Todd B Parrish
- Physical Therapy and Human Movement Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - David M Walton
- School of Physical Therapy, Western University, London, ON, Canada
| | - Sarah D Linnstaedt
- Institute for Trauma Recovery, University of North Carolina, Campus Box #7010, Chapel Hill, NC, 27599-7010, USA.
- Department of Anesthesiology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
5
|
Iannotta M, Belardo C, Trotta MC, Iannotti FA, Vitale RM, Maisto R, Boccella S, Infantino R, Ricciardi F, Mirto BF, Ferraraccio F, Panarese I, Amodeo P, Tunisi L, Cristino L, D’Amico M, di Marzo V, Luongo L, Maione S, Guida F. N-palmitoyl-D-glucosamine, a Natural Monosaccharide-Based Glycolipid, Inhibits TLR4 and Prevents LPS-Induced Inflammation and Neuropathic Pain in Mice. Int J Mol Sci 2021; 22:ijms22031491. [PMID: 33540826 PMCID: PMC7867376 DOI: 10.3390/ijms22031491] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/22/2022] Open
Abstract
Toll-like receptors (TLRs) are key receptors through which infectious and non-infectious challenges act with consequent activation of the inflammatory cascade that plays a critical function in various acute and chronic diseases, behaving as amplification and chronicization factors of the inflammatory response. Previous studies have shown that synthetic analogues of lipid A based on glucosamine with few chains of unsaturated and saturated fatty acids, bind MD-2 and inhibit TLR4 receptors. These synthetic compounds showed antagonistic activity against TLR4 activation in vitro by LPS, but little or no activity in vivo. This study aimed to show the potential use of N-palmitoyl-D-glucosamine (PGA), a bacterial molecule with structural similarity to the lipid A component of LPS, which could be useful for preventing LPS-induced tissue damage or even peripheral neuropathies. Molecular docking and molecular dynamics simulations showed that PGA stably binds MD-2 with a MD-2/(PGA)3 stoichiometry. Treatment with PGA resulted in the following effects: (i) it prevented the NF-kB activation in LPS stimulated RAW264.7 cells; (ii) it decreased LPS-induced keratitis and corneal pro-inflammatory cytokines, whilst increasing anti-inflammatory cytokines; (iii) it normalized LPS-induced miR-20a-5p and miR-106a-5p upregulation and increased miR-27a-3p levels in the inflamed corneas; (iv) it decreased allodynia in peripheral neuropathy induced by oxaliplatin or formalin, but not following spared nerve injury of the sciatic nerve (SNI); (v) it prevented the formalin- or oxaliplatin-induced myelino-axonal degeneration of sciatic nerve. SIGNIFICANCE STATEMENT We report that PGA acts as a TLR4 antagonist and this may be the basis of its potent anti-inflammatory activity. Being unique because of its potency and stability, as compared to other similar congeners, PGA can represent a tool for the optimization of new TLR4 modulating drugs directed against the cytokine storm and the chronization of inflammation.
Collapse
Affiliation(s)
- Monica Iannotta
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.I.); (C.B.); (M.C.T.); (R.M.); (S.B.); (R.I.); (F.R.); (B.F.M.); (M.D.); (L.L.)
| | - Carmela Belardo
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.I.); (C.B.); (M.C.T.); (R.M.); (S.B.); (R.I.); (F.R.); (B.F.M.); (M.D.); (L.L.)
| | - Maria Consiglia Trotta
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.I.); (C.B.); (M.C.T.); (R.M.); (S.B.); (R.I.); (F.R.); (B.F.M.); (M.D.); (L.L.)
| | - Fabio Arturo Iannotti
- Institute of Biomolecular Chemistry (ICB) of National Research Council (CNR), 80078 Pozzuoli, Italy; (F.A.I.); (R.M.V.); (P.A.); (L.T.); (L.C.); (V.d.M.)
| | - Rosa Maria Vitale
- Institute of Biomolecular Chemistry (ICB) of National Research Council (CNR), 80078 Pozzuoli, Italy; (F.A.I.); (R.M.V.); (P.A.); (L.T.); (L.C.); (V.d.M.)
| | - Rosa Maisto
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.I.); (C.B.); (M.C.T.); (R.M.); (S.B.); (R.I.); (F.R.); (B.F.M.); (M.D.); (L.L.)
| | - Serena Boccella
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.I.); (C.B.); (M.C.T.); (R.M.); (S.B.); (R.I.); (F.R.); (B.F.M.); (M.D.); (L.L.)
| | - Rosmara Infantino
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.I.); (C.B.); (M.C.T.); (R.M.); (S.B.); (R.I.); (F.R.); (B.F.M.); (M.D.); (L.L.)
| | - Flavia Ricciardi
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.I.); (C.B.); (M.C.T.); (R.M.); (S.B.); (R.I.); (F.R.); (B.F.M.); (M.D.); (L.L.)
| | - Benito Fabio Mirto
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.I.); (C.B.); (M.C.T.); (R.M.); (S.B.); (R.I.); (F.R.); (B.F.M.); (M.D.); (L.L.)
| | - Franca Ferraraccio
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.F.); (I.P.)
| | - Iacopo Panarese
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (F.F.); (I.P.)
| | - Pietro Amodeo
- Institute of Biomolecular Chemistry (ICB) of National Research Council (CNR), 80078 Pozzuoli, Italy; (F.A.I.); (R.M.V.); (P.A.); (L.T.); (L.C.); (V.d.M.)
| | - Lea Tunisi
- Institute of Biomolecular Chemistry (ICB) of National Research Council (CNR), 80078 Pozzuoli, Italy; (F.A.I.); (R.M.V.); (P.A.); (L.T.); (L.C.); (V.d.M.)
| | - Luigia Cristino
- Institute of Biomolecular Chemistry (ICB) of National Research Council (CNR), 80078 Pozzuoli, Italy; (F.A.I.); (R.M.V.); (P.A.); (L.T.); (L.C.); (V.d.M.)
| | - Michele D’Amico
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.I.); (C.B.); (M.C.T.); (R.M.); (S.B.); (R.I.); (F.R.); (B.F.M.); (M.D.); (L.L.)
| | - Vincenzo di Marzo
- Institute of Biomolecular Chemistry (ICB) of National Research Council (CNR), 80078 Pozzuoli, Italy; (F.A.I.); (R.M.V.); (P.A.); (L.T.); (L.C.); (V.d.M.)
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Faculty of Medicine and Faculty of Agriculture and Food Science, Universitè Laval, Quebec City, QC G1V 0A6, Canada
| | - Livio Luongo
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.I.); (C.B.); (M.C.T.); (R.M.); (S.B.); (R.I.); (F.R.); (B.F.M.); (M.D.); (L.L.)
- I.R.C.S.S., Neuromed, 86077 Pozzilli, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.I.); (C.B.); (M.C.T.); (R.M.); (S.B.); (R.I.); (F.R.); (B.F.M.); (M.D.); (L.L.)
- I.R.C.S.S., Neuromed, 86077 Pozzilli, Italy
- Correspondence: (S.M.); (F.G.); Tel.: +39-0815667658 (F.G.)
| | - Francesca Guida
- Department of Experimental Medicine, Pharmacology Division, University of Campania “L. Vanvitelli”, 80138 Naples, Italy; (M.I.); (C.B.); (M.C.T.); (R.M.); (S.B.); (R.I.); (F.R.); (B.F.M.); (M.D.); (L.L.)
- Correspondence: (S.M.); (F.G.); Tel.: +39-0815667658 (F.G.)
| |
Collapse
|
6
|
Micheli L, Vasarri M, Barletta E, Lucarini E, Ghelardini C, Degl’Innocenti D, Di Cesare Mannelli L. Efficacy of Posidonia oceanica Extract against Inflammatory Pain: In Vivo Studies in Mice. Mar Drugs 2021; 19:md19020048. [PMID: 33494253 PMCID: PMC7909763 DOI: 10.3390/md19020048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 12/27/2022] Open
Abstract
Posidonia oceanica (L.) Delile is traditionally used for its beneficial properties. Recently, promising antioxidant and anti-inflammatory biological properties emerged through studying the in vitro activity of the ethanolic leaves extract (POE). The present study aims to investigate the anti-inflammatory and analgesic role of POE in mice. Inflammatory pain was modeled in CD-1 mice by the intraplantar injection of carrageenan, interleukin IL-1β and formalin. Pain threshold was measured by von Frey and paw pressure tests. Nociceptive pain was studied by the hot-plate test. POE (10–100 mg kg−1) was administered per os. The paw soft tissue of carrageenan-treated animals was analyzed to measure anti-inflammatory and antioxidant effects. POE exerted a dose-dependent, acute anti-inflammatory effect able to counteract carrageenan-induced pain and paw oedema. Similar anti-hyperalgesic and anti-allodynic results were obtained when inflammation was induced by IL-1β. In the formalin test, the pre-treatment with POE significantly reduced the nocifensive behavior. Moreover, POE was able to evoke an analgesic effect in naïve animals. Ex vivo, POE reduced the myeloperoxidase activity as well as TNF-α and IL-1β levels; further antioxidant properties were highlighted as a reduction in NO concentration. POE is the candidate for a new valid strategy against inflammation and pain.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA)‐Pharmacology and Toxicology Section, University of Florence, Viale Gaetano Pieraccini, 6, 50139 Florence, Italy; (L.M.); (E.L.); (C.G.)
| | - Marzia Vasarri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.V.); (E.B.); (D.D.)
| | - Emanuela Barletta
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.V.); (E.B.); (D.D.)
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA)‐Pharmacology and Toxicology Section, University of Florence, Viale Gaetano Pieraccini, 6, 50139 Florence, Italy; (L.M.); (E.L.); (C.G.)
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA)‐Pharmacology and Toxicology Section, University of Florence, Viale Gaetano Pieraccini, 6, 50139 Florence, Italy; (L.M.); (E.L.); (C.G.)
| | - Donatella Degl’Innocenti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134 Florence, Italy; (M.V.); (E.B.); (D.D.)
- Interuniversity Center of Marine Biology and Applied Ecology “G. Bacci” (CIBM), Viale N. Sauro 4, 57128 Livorno, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA)‐Pharmacology and Toxicology Section, University of Florence, Viale Gaetano Pieraccini, 6, 50139 Florence, Italy; (L.M.); (E.L.); (C.G.)
- Correspondence:
| |
Collapse
|
7
|
Anand KJS, Roue JM, Rovnaghi CR, Marx W, Bornmann L. Historical roots of pain management in infants: A bibliometric analysis using reference publication year spectroscopy. PAEDIATRIC & NEONATAL PAIN 2020; 2:22-32. [PMID: 35548591 PMCID: PMC8975229 DOI: 10.1002/pne2.12035] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 07/03/2020] [Accepted: 07/03/2020] [Indexed: 11/08/2022]
Abstract
Retrospective evaluations of the historical role of previously published research are often fraught with subjective bias and misrepresentation, which leads to contested scientific claims. This paper investigates the historical roots of infant pain management using novel quantitative methods to identify the published literature and evaluate its relative importance. A bibliometric analysis named “reference publication year spectroscopy” (RPYS), was performed using the program CitedReferencesExplorer (CRExplorer) to avoid the subjectivity associated with comparative evaluations of individual research studies. Web of Science (WoS) search queries on infant‐related synonyms, pain‐related synonyms, and analgesia or anesthesia‐related synonyms were combined using the Boolean operator “AND,” to identify all publications related to pain management in infants. The RPYS analyses were based on 8697 papers in our publication set containing the citations for 86268 references. Selected cited publications were associated with peak citation years in 1951, 1954, 1957, 1965, 1987, 1990, 1997, 1999, and 2000. Subsequent analyses suggested that research on infant pain management made rapid progress during 1982‐1992. Landmark publications were defined as those belonging to the top 10% of the most frequently referenced publications for longer than 25 years. Through this analysis, we identified and ranked 24 landmark publications to illustrate the historical background and early research on infant pain management. From the first‐ever application of RPYS (an objective, reproducible approach to study the early history of any scholarly activity) to pain research, infant pain management appears rooted in the scientific rationale for neonatal pain perception, randomized trials of opioid anesthesia/analgesia, and studies describing the facial expressions and crying activity following heel‐lance procedures in newborns.
Collapse
Affiliation(s)
- Kanwaljeet J S Anand
- Department of Pediatrics Stanford University School of Medicine Palo Alto CA USA.,Pain/Stress Neurobiology Lab Maternal & Child Health Research Institute Stanford University School of Medicine Palo Alto CA USA
| | - Jean-Michel Roue
- Neonatal & Pediatric Intensive Care Unit Brest University Hospital University of Western Brittany Brest France
| | - Cynthia R Rovnaghi
- Pain/Stress Neurobiology Lab Maternal & Child Health Research Institute Stanford University School of Medicine Palo Alto CA USA
| | - Werner Marx
- Max Planck Institute for Solid State Research Stuttgart Germany
| | - Lutz Bornmann
- Division for Science and Innovation Studies Administrative Headquarters The Max Planck Society Munich Germany
| |
Collapse
|
8
|
Linciano P, Citti C, Luongo L, Belardo C, Maione S, Vandelli MA, Forni F, Gigli G, Laganà A, Montone CM, Cannazza G. Isolation of a High-Affinity Cannabinoid for the Human CB1 Receptor from a Medicinal Cannabis sativa Variety: Δ 9-Tetrahydrocannabutol, the Butyl Homologue of Δ 9-Tetrahydrocannabinol. JOURNAL OF NATURAL PRODUCTS 2020; 83:88-98. [PMID: 31891265 DOI: 10.1021/acs.jnatprod.9b00876] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The butyl homologues of Δ9-tetrahydrocannabinol, Δ9-tetrahydrocannabutol (Δ9-THCB), and cannabidiol, cannabidibutol (CBDB), were isolated from a medicinal Cannabis sativa variety (FM2) inflorescence. Appropriate spectroscopic and spectrometric characterization, including NMR, UV, IR, ECD, and HRMS, was carried out on both cannabinoids. The chemical structures and absolute configurations of the isolated cannabinoids were confirmed by comparison with the spectroscopic data of the respective compounds obtained by stereoselective synthesis. The butyl homologue of Δ9-THC, Δ9-THCB, showed an affinity for the human CB1 (Ki = 15 nM) and CB2 receptors (Ki = 51 nM) comparable to that of (-)-trans-Δ9-THC. Docking studies suggested the key bonds responsible for THC-like binding affinity for the CB1 receptor. The formalin test in vivo was performed on Δ9-THCB in order to reveal possible analgesic and anti-inflammatory properties. The tetrad test in mice showed a partial agonistic activity of Δ9-THCB toward the CB1 receptor.
Collapse
MESH Headings
- Analgesics/pharmacology
- Animals
- Cannabidiol/chemistry
- Cannabinoids/chemistry
- Cannabinoids/isolation & purification
- Cannabis/chemistry
- Dronabinol/chemistry
- Dronabinol/isolation & purification
- Humans
- Medical Marijuana
- Mice
- Molecular Structure
- Receptor, Cannabinoid, CB1/chemistry
- Receptor, Cannabinoid, CB1/isolation & purification
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/chemistry
- Receptor, Cannabinoid, CB2/metabolism
Collapse
Affiliation(s)
- Pasquale Linciano
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Campi 103 , 41125 Modena , Italy
| | - Cinzia Citti
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Campi 103 , 41125 Modena , Italy
- Mediteknology s.r.l. , Via Arnesano , 73100 Lecce , Italy
- CNR NANOTEC , Campus Ecotekne, Via Monteroni , 73100 Lecce , Italy
| | - Livio Luongo
- Department of Experimental Medicine, Division of Pharmacology , Università della Campania "L. Vanvitelli" , Via Santa Maria di Costantinopoli 16 , 80138 Naples , Italy
| | - Carmela Belardo
- Department of Experimental Medicine, Division of Pharmacology , Università della Campania "L. Vanvitelli" , Via Santa Maria di Costantinopoli 16 , 80138 Naples , Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Division of Pharmacology , Università della Campania "L. Vanvitelli" , Via Santa Maria di Costantinopoli 16 , 80138 Naples , Italy
| | - Maria Angela Vandelli
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Campi 103 , 41125 Modena , Italy
| | - Flavio Forni
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Campi 103 , 41125 Modena , Italy
| | - Giuseppe Gigli
- CNR NANOTEC , Campus Ecotekne, Via Monteroni , 73100 Lecce , Italy
| | - Aldo Laganà
- CNR NANOTEC , Campus Ecotekne, Via Monteroni , 73100 Lecce , Italy
- Department of Chemistry , Sapienza University of Rome , Piazzale Aldo Moro 5 , 00185 Rome , Italy
| | - Carmela Maria Montone
- Department of Chemistry , Sapienza University of Rome , Piazzale Aldo Moro 5 , 00185 Rome , Italy
| | - Giuseppe Cannazza
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Campi 103 , 41125 Modena , Italy
- CNR NANOTEC , Campus Ecotekne, Via Monteroni , 73100 Lecce , Italy
| |
Collapse
|
9
|
Mascarenhas CJ, Liu R, Barr GA. Effects of plant-derived analgesic compounds sinomenine and salvinorin A in infant rats. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2020; 18:174-180. [PMID: 31992510 DOI: 10.1016/j.joim.2020.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 07/03/2019] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Premature and ill neonates undergo painful but medically necessary procedures while hospitalized. Although opiate drugs are administered as analgesics, problems associated with their side effects, tolerance, and potential dependence necessitate research into alternative pain-relieving medications. Here we test two plant-derived compounds in infant rats: sinomenine, which targets the Mas-related G-protein-coupled receptor member X2 opioid receptor; and salvinorin A, which is a κ opioid receptor agonist. In adult animals both sinomenine and salvinorin A are analgesic, but neither has been tested in infants. METHODS We used the formalin and thermal plantar tests in rats 7 and 21 days of age (PN7 and PN21) for behavioral signs of pain. In addition, brain sections were stained using Fos immunohistochemistry to examine patterns of brain activation in the midbrain periaqueductal gray and the paraventricular nucleus of the hypothalamus. RESULTS Sinomenine was analgesic in both the formalin and thermal tests on animals 21 days of age. At PN7 only the highest dose elevated response latencies in the thermal test and there were no effects of sinomenine in the formalin test. Analysis of Fos expression in the sinomenine-treated animals showed no drug effect, in contrast to the behavioral results. Salvinorin A was analgesic in the formalin test only at the highest dose at 21 days of age but not in the thermal test at either age. CONCLUSION The increased modest effectiveness of sinomenine in older animals and the minimum salvinorin A drug effect suggest that the compounds act on sites that develop during the preweaning period (sinomenine) or after weaning (salvinorin A).
Collapse
Affiliation(s)
- Conrad J Mascarenhas
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Renyu Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gordon A Barr
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Anesthesiology and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Butkevich IP, Mikhailenko VA, Vershinina EA, Barr GA. Differences Between the Prenatal Effects of Fluoxetine or Buspirone Alone or in Combination on Pain and Affective Behaviors in Prenatally Stressed Male and Female Rats. Front Behav Neurosci 2019; 13:125. [PMID: 31244623 PMCID: PMC6579839 DOI: 10.3389/fnbeh.2019.00125] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/22/2019] [Indexed: 01/06/2023] Open
Abstract
The selective serotonin reuptake inhibitor fluoxetine and the 5-HT1A receptor agonist buspirone are used to treat depression and anxiety. Previously we demonstrated that chronic stress during pregnancy (prenatal stress) in rats, used as a model of maternal depression risk, increased inflammatory pain and depressive-like behavior in the offspring; buspirone injected to pregnant dams was protective. Clinically, the addition of buspirone to fluoxetine increases the latter’s efficacy in treating depression in patients. Here, we investigated the influence of repeated prenatal injections of fluoxetine, buspirone or their combination on pain- and depressive-like behaviors in prenatally stressed young male and female rats. Prenatal stress augmented depressive-like behavior and both thermal and inflammatory pain (formalin test), replicating our prior findings, and increased basal levels of corticosterone in the blood plasma. Both drugs and their combination reduced the effects of prenatal stress on thermal pain and depressive-like behavior independently of sex. The combination of fluoxetine and buspirone, compared with fluoxetine, was more antinociceptive in the hot plate test in both sexes, and when compared with buspirone, was more antinociceptive only in males. A detailed study of the time-course of formalin-induced pain showed a nuanced effect of these drugs that was sex-dependent. The combination of the two drugs was less effective in females than males during the initial acute phase of nociceptive behavior in flexing + shaking behaviors, whereas that combination was more effective than fluoxetine alone in the first acute phase of licking behavior in females. The antinociceptive effect of buspirone dominated that of the drug combination and of fluoxetine alone, especially during the interphase of the formalin test in both sexes for both flexing + shaking and licking, suggesting a more effective prenatal action of buspirone on the development of a descending serotonergic inhibitory system modulating pain in the spinal cord dorsal horn neurons. Our results indicate that inflammatory pain-like responses integrated at the spinal level in males were more vulnerable to prenatal stress than females. In licking, the antinociceptive effect of fluoxetine and drug combination in the interphase was more in males than females. The data underscore the importance of considering sexual dimorphism when using drug therapy.
Collapse
Affiliation(s)
- Irina P Butkevich
- Laboratory of Ontogenesis of the Nervous System, I.P. Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, Russia.,Department of Normal Physiology, State Pediatric Medical University, St. Petersburg, Russia
| | - Viktor A Mikhailenko
- Laboratory of Ontogenesis of the Nervous System, I.P. Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Elena A Vershinina
- Department of Information Technologies and Mathematical Modeling, I.P. Pavlov Institute of Physiology, Russian Academy of Sciences, St. Petersburg, Russia
| | - Gordon A Barr
- Department of Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
11
|
Fu H, Fang P, Zhou HY, Zhou J, Yu XW, Ni M, Zheng JY, Jin Y, Chen JG, Wang F, Hu ZL. Acid-sensing ion channels in trigeminal ganglion neurons innervating the orofacial region contribute to orofacial inflammatory pain. Clin Exp Pharmacol Physiol 2016; 43:193-202. [PMID: 26510178 DOI: 10.1111/1440-1681.12510] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 10/21/2015] [Accepted: 10/22/2015] [Indexed: 12/31/2022]
Abstract
Orofacial pain is a common clinical symptom that is accompanied by tooth pain, migraine and gingivitis. Accumulating evidence suggests that acid-sensing ion channels (ASICs), especially ASIC3, can profoundly affect the physiological properties of nociception in peripheral sensory neurons. The aim of this study is to examine the contribution of ASICs in trigeminal ganglion (TG) neurons to orofacial inflammatory pain. A Western blot (WB), immunofluorescence assay of labelled trigeminal ganglion neurons, orofacial formalin test, cell preparation and electrophysiological experiments are performed. This study demonstrated that ASIC1, ASIC2a and ASIC3 are highly expressed in TG neurons innervating the orofacial region of rats. The amplitude of ASIC currents in these neurons increased 119.72% (for ASIC1-like current) and 230.59% (for ASIC3-like current) in the formalin-induced orofacial inflammatory pain model. In addition, WB and immunofluorescence assay demonstrated a significantly augmented expression of ASICs in orofacial TG neurons during orofacial inflammation compared with the control group. The relative protein density of ASIC1, ASIC2a and ASIC3 also increased 58.82 ± 8.92%, 45.30 ± 11.42% and 55.32 ± 14.71%, respectively, compared with the control group. Furthermore, pharmacological blockade of ASICs and genetic deletion of ASIC1 attenuated the inflammation response. These findings indicate that peripheral inflammation can induce the upregulation of ASICs in TG neurons, causing orofacial inflammatory pain. Additionally, the specific inhibitor of ASICs may have a significant analgesic effect on orofacial inflammatory pain.
Collapse
Affiliation(s)
- Hui Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei.,Department of Pharmacology, Guangdong Medical College, Dongguan, China
| | - Peng Fang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Hai-Yun Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Jun Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Xiao-Wei Yu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Ming Ni
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - Jie-Yan Zheng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei
| | - You Jin
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei.,The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei.,The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei
| | - Jian-Guo Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei.,The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei.,The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei
| | - Fang Wang
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei.,The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei.,The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei
| | - Zhuang-Li Hu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei.,The Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, Hubei.,The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, Hubei
| |
Collapse
|
12
|
Aiello F, Badolato M, Pessina F, Sticozzi C, Maestrini V, Aldinucci C, Luongo L, Guida F, Ligresti A, Artese A, Allarà M, Costa G, Frosini M, Schiano Moriello A, De Petrocellis L, Valacchi G, Alcaro S, Maione S, Di Marzo V, Corelli F, Brizzi A. Design and Synthesis of New Transient Receptor Potential Vanilloid Type-1 (TRPV1) Channel Modulators: Identification, Molecular Modeling Analysis, and Pharmacological Characterization of the N-(4-Hydroxy-3-methoxybenzyl)-4-(thiophen-2-yl)butanamide, a Small Molecule Endowed with Agonist TRPV1 Activity and Protective Effects against Oxidative Stress. ACS Chem Neurosci 2016; 7:737-48. [PMID: 26942555 DOI: 10.1021/acschemneuro.5b00333] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
4-(Thiophen-2-yl)butanoic acid was identified as a cyclic substitute of the unsaturated alkyl chain of the natural ligand, capsaicin. Accordingly, a new class of amides was synthesized in good yield and high purity and their molecular recognition against the target was investigated by means of docking experiments followed by molecular dynamics simulations, in order to rationalize their geometrical and thermodynamic profiles. The pharmacological properties of these new compounds were expressed as activation (EC50) and desensitization (IC50) potencies. Several compounds were found to activate TRPV1 channels, and in particular, derivatives 1 and 10 behaved as TRPV1 agonists endowed with good efficacy as compared to capsaicin. The most promising compound 1 was also evaluated for its protective role against oxidative stress on keratinocytes and differentiated human neuroblastoma cell lines expressing the TRPV1 receptor as well as for its cytotoxicity and analgesic activity in vivo.
Collapse
Affiliation(s)
- Francesca Aiello
- Dipartimento
di Farmacia e Scienza della Salute e della Nutrizione, Università della Calabria, Edificio Polifunzionale, 87036 Arcavacata di Rende, Cosenza, Italy
| | - Mariateresa Badolato
- Dipartimento
di Farmacia e Scienza della Salute e della Nutrizione, Università della Calabria, Edificio Polifunzionale, 87036 Arcavacata di Rende, Cosenza, Italy
| | | | - Claudia Sticozzi
- Dipartimento
Scienza della Vita e Biotecnologie, Università degli Studi di Ferrara, Via L. Borsari 46, 44121 Ferrara, Italy
| | | | | | - Livio Luongo
- Dipartimento
di Medicina Sperimentale, Sezione di Farmacologia “L. Donatelli”, Seconda Università di Napoli, 80138 Napoli, Italy
| | - Francesca Guida
- Dipartimento
di Medicina Sperimentale, Sezione di Farmacologia “L. Donatelli”, Seconda Università di Napoli, 80138 Napoli, Italy
| | - Alessia Ligresti
- Istituto
di Chimica Biomolecolare, Endocannabinoid Research Group, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy
| | - Anna Artese
- Dipartimento
di Scienze della Salute, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Marco Allarà
- Istituto
di Chimica Biomolecolare, Endocannabinoid Research Group, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy
| | - Giosué Costa
- Dipartimento
di Scienze della Salute, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | | | - Aniello Schiano Moriello
- Istituto
di Chimica Biomolecolare, Endocannabinoid Research Group, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy
| | - Luciano De Petrocellis
- Istituto
di Chimica Biomolecolare, Endocannabinoid Research Group, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy
| | - Giuseppe Valacchi
- Dipartimento
Scienza della Vita e Biotecnologie, Università degli Studi di Ferrara, Via L. Borsari 46, 44121 Ferrara, Italy
| | - Stefano Alcaro
- Dipartimento
di Scienze della Salute, Università degli Studi “Magna Graecia” di Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Sabatino Maione
- Dipartimento
di Medicina Sperimentale, Sezione di Farmacologia “L. Donatelli”, Seconda Università di Napoli, 80138 Napoli, Italy
| | - Vincenzo Di Marzo
- Istituto
di Chimica Biomolecolare, Endocannabinoid Research Group, Consiglio Nazionale delle Ricerche, Via Campi Flegrei 34, 80078 Pozzuoli, Napoli, Italy
| | | | | |
Collapse
|
13
|
Co-administration of morphine and gabapentin leads to dose dependent synergistic effects in a rat model of postoperative pain. Eur J Pharm Sci 2015; 82:97-105. [PMID: 26610393 DOI: 10.1016/j.ejps.2015.11.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/30/2015] [Accepted: 11/18/2015] [Indexed: 12/21/2022]
Abstract
Despite much evidence that combination of morphine and gabapentin can be beneficial for managing postoperative pain, the nature of the pharmacological interaction of the two drugs remains unclear. The aim of this study was to assess the interaction of morphine and gabapentin in range of different dose combinations and investigate whether co-administration leads to synergistic effects in a preclinical model of postoperative pain. The pharmacodynamic effects of morphine (1, 3 and 7mg/kg), gabapentin (10, 30 and 100mg/kg) or their combination (9 combinations in total) were evaluated in the rat plantar incision model using an electronic von Frey device. The percentage of maximum possible effect (%MPE) and the area under the response curve (AUC) were used for evaluation of the antihyperalgesic effects of the drugs. Identification of synergistic interactions was based on Loewe additivity response surface analyses. The combination of morphine and gabapentin resulted in synergistic antihyperalgesic effects in a preclinical model of postoperative pain. The synergistic interactions were found to be dose dependent and the increase in observed response compared to the theoretical additive response ranged between 26 and 58% for the synergistic doses. The finding of dose-dependent synergistic effects highlights that choosing the right dose-dose combination is of importance in postoperative pain therapy. Our results indicate benefit of high doses of gabapentin as adjuvant to morphine. If these findings translate to humans, they might have important implications for the treatment of pain in postoperative patients.
Collapse
|
14
|
Abstract
Safety signals provide "relief" through predicting the absence of an aversive event. At issue is whether these signals also act as instrumental reinforcers. Four experiments were conducted using a free-operant lever-press avoidance paradigm in which each press avoided shock and was followed by the presentation of a 5-sec auditory safety signal. When given a choice between two levers in Experiment 1, both avoiding shock, rats preferentially responded on the lever that produced the safety signal as feedback, even when footshock was omitted. Following avoidance training with a single lever in Experiment 2, removal of the signal led to a decrease in avoidance responses and an increase in responses during the safety period normally denoted by the signal. These behavioral changes demonstrate the dual conditioned reinforcing and fear inhibiting properties of the safety signal. The associative processes that support the reinforcing properties of a safety signal were tested using a novel revaluation procedure. Prior experience of systemic morphine during safety signal presentations resulted in an increased rate of avoidance responses to produce the safety signal during a drug-free extinction test, a finding not seen with d-amphetamine in Experiment 3. Morphine revaluation of the safety signal was repeated in Experiment 4 followed by a drug-free extinction test in which responses did not produce the signal for the first 10 min of the session. Instrumental avoidance in the absence of the signal was shown to be insensitive to prior signal revaluation, suggesting that the signal reinforces free-operant avoidance behavior through a habit-like mechanism.
Collapse
|
15
|
Free-operant avoidance behavior by rats after reinforcer revaluation using opioid agonists and D-amphetamine. J Neurosci 2014; 34:6286-93. [PMID: 24790199 DOI: 10.1523/jneurosci.4146-13.2014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The associative processes that support free-operant instrumental avoidance behavior are still unknown. We used a revaluation procedure to determine whether the performance of an avoidance response is sensitive to the current value of the aversive, negative reinforcer. Rats were trained on an unsignaled, free-operant lever press avoidance paradigm in which each response avoided or escaped shock and produced a 5 s feedback stimulus. The revaluation procedure consisted of noncontingent presentations of the shock in the absence of the lever either paired or unpaired with systemic morphine and in a different cohort with systemic d-amphetamine. Rats were then tested drug free during an extinction test. In both the d-amphetamine and morphine groups, pairing of the drug and shock decreased subsequent avoidance responding during the extinction test, suggesting that avoidance behavior was sensitive to the current incentive value of the aversive negative reinforcer. Experiment 2 used central infusions of D-Ala(2), NMe-Phe(4), Gly-ol(5)]-enkephalin (DAMGO), a mu-opioid receptor agonist, in the periacqueductal gray and nucleus accumbens shell to revalue the shock. Infusions of DAMGO in both regions replicated the effects seen with systemic morphine. These results are the first to demonstrate the impact of revaluation of an aversive reinforcer on avoidance behavior using pharmacological agents, thereby providing potential therapeutic targets for the treatment of avoidance behavior symptomatic of anxiety disorders.
Collapse
|
16
|
Behavioral effects of perinatal opioid exposure. Life Sci 2014; 104:1-8. [PMID: 24746901 DOI: 10.1016/j.lfs.2014.04.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 04/01/2014] [Accepted: 04/05/2014] [Indexed: 01/15/2023]
Abstract
Opioids are among the world's oldest known drugs used mostly for pain relief, but recreational use is also widespread. A particularly important problem is opioid exposure in females, as their offspring can also be affected. Adverse intrauterine and postnatal environments can affect offspring development and may lead to various disabilities later in life. It is clear that repetitive painful experiences, such as randomly occurring invasive procedures during neonatal intensive care, can permanently alter neuronal and synaptic organization and therefore later behavior. At the same time, analgesic drugs can also be harmful, inducing neuronal apoptosis or withdrawal symptoms in the neonate and behavioral alterations in adulthood. Hence, risk-benefit ratios should be taken into consideration when pain relief is required during pregnancy or in neonates. Recreational use of opioids can also alter many aspects of life. Intrauterine opioid exposure has many toxic effects, inducing poor pregnancy outcomes due to underdevelopment, but it is believed that later negative consequences are more related to environmental factors such as a chaotic lifestyle and inadequate prenatal care. One of the crucial components is maternal care, which changes profoundly in addicted mothers. In substance-dependent mothers, pre- and postnatal care has special importance, and controlled treatment with a synthetic opioid (e.g., methadone) could be beneficial. We aimed to summarize and compare human and rodent data, as it is important to close the gap between scientific knowledge and societal policies. Special emphasis is given to gender differences in the sensitivity of offspring to perinatal opioid exposure.
Collapse
|
17
|
Maud P, Thavarak O, Cédrick L, Michèle B, Vincent B, Olivier P, Régis B. Evidence for the use of isoflurane as a replacement for chloral hydrate anesthesia in experimental stroke: an ethical issue. BIOMED RESEARCH INTERNATIONAL 2014; 2014:802539. [PMID: 24719888 PMCID: PMC3955691 DOI: 10.1155/2014/802539] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/07/2014] [Indexed: 01/10/2023]
Abstract
Since an ethical issue has been raised regarding the use of the well-known anesthetic agent chloral hydrate, owing to its mutagenic and carcinogenic effects in animals, attention of neuroscientists has turned to finding out an alternative agent able to meet not only potency, safety, and analgesic efficacy, but also reduced neuroprotective effect for stroke research. The aim of this study was to compare the potential of chloral hydrate and isoflurane for both modulating the action of the experimental neuroprotectant MK801 and exerting analgesia. After middle cerebral artery occlusion in rats, no difference was observed in 24 h survival rate, success of ischemia, or infarct volume reduction between both anesthetics. However, isoflurane exerted a more pronounced analgesic effect than chloral hydrate as evidenced by formalin test 3 hours after anesthesia onset, thus encouraging the use of isoflurane in experimental stroke models.
Collapse
Affiliation(s)
- Pétrault Maud
- EA 1046-Département de Pharmacologie Médicale, Faculté de Médecine, CHU Lille, 1 Place de Verdun, 59045 Lille Cedex, France
- UDSL, 59000 Lille, France
| | - Ouk Thavarak
- EA 1046-Département de Pharmacologie Médicale, Faculté de Médecine, CHU Lille, 1 Place de Verdun, 59045 Lille Cedex, France
- UDSL, 59000 Lille, France
| | - Lachaud Cédrick
- EA 1046-Département de Pharmacologie Médicale, Faculté de Médecine, CHU Lille, 1 Place de Verdun, 59045 Lille Cedex, France
- UDSL, 59000 Lille, France
| | - Bastide Michèle
- EA 1046-Département de Pharmacologie Médicale, Faculté de Médecine, CHU Lille, 1 Place de Verdun, 59045 Lille Cedex, France
- IUT A, Université de Lille 1, 59653 Villeneuve d'Ascq Cedex, France
| | - Bérézowski Vincent
- EA 1046-Département de Pharmacologie Médicale, Faculté de Médecine, CHU Lille, 1 Place de Verdun, 59045 Lille Cedex, France
- Université d'Artois, 62307 Lens, France
| | - Pétrault Olivier
- EA 1046-Département de Pharmacologie Médicale, Faculté de Médecine, CHU Lille, 1 Place de Verdun, 59045 Lille Cedex, France
- Université d'Artois, 62307 Lens, France
| | - Bordet Régis
- EA 1046-Département de Pharmacologie Médicale, Faculté de Médecine, CHU Lille, 1 Place de Verdun, 59045 Lille Cedex, France
- UDSL, 59000 Lille, France
| |
Collapse
|
18
|
Luongo L, Costa B, D'Agostino B, Guida F, Comelli F, Gatta L, Matteis M, Sullo N, De Petrocellis L, de Novellis V, Maione S, Di Marzo V. Palvanil, a non-pungent capsaicin analogue, inhibits inflammatory and neuropathic pain with little effects on bronchopulmonary function and body temperature. Pharmacol Res 2012; 66:243-50. [PMID: 22634607 DOI: 10.1016/j.phrs.2012.05.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 05/15/2012] [Accepted: 05/16/2012] [Indexed: 10/28/2022]
Abstract
N-Palmitoyl-vanillamide (palvanil) is a non-pungent capsaicinoid, found in low amounts in Capsicum and shown to rapidly desensitize transient receptor potential vanilloid type-1 (TRPV1) channels to the action of capsaicin and to exert analgesic effects after local administration. We have investigated here if systemic administration of palvanil to mice causes two typical adverse events of TRPV1 agonists, i.e. profound changes in body temperature and bronchoconstriction, and if it can still produce effective inhibition of inflammatory and chronic pain in different experimental models. Varying doses of palvanil were tested subcutaneously and acutely on body temperature in vivo or, or as a bolus, on bronchopulmunary function ex vivo, in comparison with capsaicin. Intraperitoneal palvanil was also tested against formalin-induced nocifensive behavior and carrageenan-induced oedema and thermal hyperalgesia, acutely, and against mechanical allodynia and thermal hyperalgesia in mice with spared nerve injury (SNI) of the sciatic nerve, after repeated administration over 7 days from SNI. Palvanil, at therapeutically relevant doses, produced significantly less hypothermia and bronchoconstriction than capsaicin. Palvanil (0.5-2.5 mg/kg) abolished formalin-induced nocifensive behavior and strongly attenuated SNI-induced mechanical allodynia and thermal hyperalgesia and carrageenan-induced oedema and thermal hyperalgesia. Systemic administration of the non-pungent capsaicinoid, palvanil, produces, at least in mice, much less of those side effects typical of TRPV1 agonists (hypothermia and bronchoconstriction), whilst being very effective at reducing pain and oedema. Thus, palvanil might be developed further as a novel pharmacological treatment for chronic abnormal pain.
Collapse
Affiliation(s)
- Livio Luongo
- Department of Experimental Medicine-Division of Pharmacology L. Donatelli, Second University of Naples, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Butkevich IP, Mikhailenko VA, Vershinina EA, Otellin VA, Aloisi AM. Buspirone before prenatal stress protects against adverse effects of stress on emotional and inflammatory pain-related behaviors in infant rats: age and sex differences. Brain Res 2011; 1419:76-84. [PMID: 21937026 DOI: 10.1016/j.brainres.2011.08.068] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 08/23/2011] [Accepted: 08/27/2011] [Indexed: 12/29/2022]
Abstract
Prenatal stress strengthens tonic pain and provokes depression. The serotoninergic system is involved in these processes. We recently showed that maternal buspirone, a 5-HT1A receptor agonist, protects against the adverse effects of in utero stress on depression and pain in adult rat offspring. Using a similar maternal treatment with buspirone, we focus here on the infant stage, which is important for the correction of prenatal abnormalities. Maternal buspirone before restraint stress during the last week of pregnancy decreased the time of immobility in the forced swim test in the infant offspring. Prenatal stress increased formalin-induced pain in the second part of the time-course of the response to formalin in males of middle infancy but in the first part of the response in males of late infancy. The effect was reversed by maternal buspirone. Pain dominated in males of both middle and late infancy but the time-course of formalin pain in infant females revealed a slower development of the processes. The results show that the time-course of formalin-induced pain in infant rats reacts to prenatal stress in an age-dependent and sexually dimorphic manner. Our finding of opposite influences of prenatal stress and buspirone before prenatal stress on formalin-induced pain during the interphase indicates that functional maturity of the descending serotonergic inhibitory system occurs in late infancy males (11-day-olds), and 5-HT1A receptors participate in this process. The data provide evidence that maternal treatment with buspirone prior to stress during pregnancy alleviates depression-like and tonic pain-related behaviors in the infant offspring.
Collapse
Affiliation(s)
- Irina P Butkevich
- Laboratory of Ontogeny of the Nervous System, Pavlov Institute of Physiology, Russian Academy of Sciences, Nab. Makarova 6, St. Petersburg 199034, Russia.
| | | | | | | | | |
Collapse
|
20
|
Ultrapotent effects of salvinorin A, a hallucinogenic compound from Salvia divinorum, on LPS-stimulated murine macrophages and its anti-inflammatory action in vivo. J Mol Med (Berl) 2011; 89:891-902. [DOI: 10.1007/s00109-011-0752-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Revised: 03/04/2011] [Accepted: 03/08/2011] [Indexed: 10/18/2022]
|
21
|
De Petrocellis L, Guida F, Moriello AS, De Chiaro M, Piscitelli F, de Novellis V, Maione S, Di Marzo V. N-palmitoyl-vanillamide (palvanil) is a non-pungent analogue of capsaicin with stronger desensitizing capability against the TRPV1 receptor and anti-hyperalgesic activity. Pharmacol Res 2011; 63:294-9. [PMID: 21215315 DOI: 10.1016/j.phrs.2010.12.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 12/31/2010] [Accepted: 12/31/2010] [Indexed: 01/22/2023]
Abstract
N-acyl-vanillamide (NAVAM) analogues of the natural pungent principle of capsicum, capsaicin, were developed several years ago as potential non-pungent analgesic compounds. N-oleoyl-vanillamide (olvanil) and N-arachidonoy-vanillamide (arvanil), in particular, were described in several publications and patents to behave as potent anti-hyperalgesic compounds in experimental models of chronic and inflammatory pain, and to activate both "capsaicin receptors", i.e. the transient receptor potential of vanilloid type-1 (TRPV1) channel, and, either directly or indirectly, cannabinoid receptors of type-1. Here we report the biochemical and pharmacological characterization of a so far neglected NAVAM, N-palmitoyl-vanillamide (palvanil), and propose its possible use instead of capsaicin, as a possible topical analgesic. Palvanil exhibited a kinetics of activation of human recombinant TRPV1-mediated intracellular calcium elevation significantly slower than that of capsaicin (t(1/2)=21s and 8s, respectively at 1μM). Slow kinetics of TRPV1 agonists were previously found to be associated with stronger potencies as TRPV1 desensitizing agents, which in turn are usually associated with lower pungency and stronger anti-hyperalgesic activity. Accordingly, palvanil desensitized the human recombinant TRPV1 to the effect of capsaicin (10nM) with significantly higher potency than capsaicin (IC(50)=0.8nM and 3.8nM, respectively), this effect reaching its maximum more rapidly (50 and 250min, respectively). Palvanil was also more potent than capsaicin at desensitizing the stimulatory effect of TRPV1 by low pH together with anandamide, which mimics conditions occurring during inflammation. In the eye-wiping assay carried out in mice, palvanil was not pungent and instead caused a strong and long-lasting inhibition of capsaicin-induced eye-wiping. Finally, intraplantar palvanil inhibited the second phase of the nociceptive response to formalin in mice. In conclusion, palvanil appears to be a non-pungent analogue of capsaicin with stronger desensitizing effects on TRPV1 and hence potentially higher anti-hyperalgesic activity.
Collapse
Affiliation(s)
- Luciano De Petrocellis
- Endocannabinoid Research Group, Institute of Cybernetics - CNR, Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Gong KR, Cao FL, He Y, Gao CY, Wang DD, Li H, Zhang FK, An YY, Lin Q, Chen J. Enhanced excitatory and reduced inhibitory synaptic transmission contribute to persistent pain-induced neuronal hyper-responsiveness in anterior cingulate cortex. Neuroscience 2010; 171:1314-25. [DOI: 10.1016/j.neuroscience.2010.10.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 10/08/2010] [Accepted: 10/12/2010] [Indexed: 12/31/2022]
|
23
|
Bolognini D, Costa B, Maione S, Comelli F, Marini P, Di Marzo V, Parolaro D, Ross RA, Gauson LA, Cascio MG, Pertwee RG. The plant cannabinoid Delta9-tetrahydrocannabivarin can decrease signs of inflammation and inflammatory pain in mice. Br J Pharmacol 2010; 160:677-87. [PMID: 20590571 DOI: 10.1111/j.1476-5381.2010.00756.x] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE The phytocannabinoid, Delta(9)-tetrahydrocannabivarin (THCV), can block cannabinoid CB(1) receptors. This investigation explored its ability to activate CB(2) receptors, there being evidence that combined CB(2) activation/CB(1) blockade would ameliorate certain disorders. EXPERIMENTAL APPROACH We tested the ability of THCV to activate CB(2) receptors by determining whether: (i) it inhibited forskolin-stimulated cyclic AMP production by Chinese hamster ovary (CHO) cells transfected with human CB(2) (hCB(2)) receptors; (ii) it stimulated [(35)S]GTPgammaS binding to hCB(2) CHO cell and mouse spleen membranes; (iii) it attenuated signs of inflammation/hyperalgesia induced in mouse hind paws by intraplantar injection of carrageenan or formalin; and (iv) any such anti-inflammatory or anti-hyperalgesic effects were blocked by a CB(1) or CB(2) receptor antagonist. KEY RESULTS THCV inhibited cyclic AMP production by hCB(2) CHO cells (EC(50)= 38 nM), but not by hCB(1) or untransfected CHO cells or by hCB(2) CHO cells pre-incubated with pertussis toxin (100 ng.mL(-1)) and stimulated [(35)S]GTPgammaS binding to hCB(2) CHO and mouse spleen membranes. THCV (0.3 or 1 mg.kg(-1) i.p.) decreased carrageenan-induced oedema in a manner that seemed to be CB(2) receptor-mediated and suppressed carrageenan-induced hyperalgesia. THCV (i.p.) also decreased pain behaviour in phase 2 of the formalin test at 1 mg.kg(-1), and in both phases of this test at 5 mg.kg(-1); these effects of THCV appeared to be CB(1) and CB(2) receptor mediated. CONCLUSIONS AND IMPLICATIONS THCV can activate CB(2) receptors in vitro and decrease signs of inflammation and inflammatory pain in mice partly via CB(1) and/or CB(2) receptor activation.
Collapse
|
24
|
LaPrairie JL, Murphy AZ. Long-term impact of neonatal injury in male and female rats: Sex differences, mechanisms and clinical implications. Front Neuroendocrinol 2010; 31:193-202. [PMID: 20144647 PMCID: PMC2849925 DOI: 10.1016/j.yfrne.2010.02.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Revised: 02/01/2010] [Accepted: 02/03/2010] [Indexed: 02/07/2023]
Abstract
Over the last several decades, the relative contribution of early life events to individual disease susceptibility has been explored extensively. Only fairly recently, however, has it become evident that abnormal or excessive nociceptive activity experienced during the perinatal period may permanently alter the normal development of the CNS and influence future responses to somatosensory input. Given the significant rise in the number of premature infants receiving high-technology intensive care over the last 20 years, ex-preterm neonates may be exceedingly vulnerable to the long-term effects of repeated invasive interventions. The present review summarizes available clinical and laboratory findings on the lasting impact of exposure to noxious stimulation during early development, with a focus on the structural and functional alterations in nociceptive circuits, and its sexually dimorphic impact.
Collapse
Affiliation(s)
- Jamie L LaPrairie
- Neuroscience Institute, Georgia State University, 38 Peachtree Center Ave, 806 GCB, Atlanta, GA 30303, USA
| | | |
Collapse
|
25
|
Interrelationship between measures of pain reactions in inflammation and levels of depression in prenatally stressed rat pups. ACTA ACUST UNITED AC 2009; 40:179-84. [PMID: 20033308 DOI: 10.1007/s11055-009-9241-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 07/21/2008] [Indexed: 11/27/2022]
Abstract
The interrelationship between measures of pain reactions (number of flexion + shaking patterns) in the formalin test and the level of depression (duration of immobility) in the forced swimming (Porsolt) test was studied in prenatally stressed rat pups aged 7-8 days. Two series of experiments were performed, with different sequences of tests separated by intervals of one day. In the first series of experiments, the Porsolt test was performed first; in the second series, the formalin test was performed before forced swimming. The sequence of tests was found to have different effects on measures of pain and depression and their correlation in prenatally stressed and unstressed rat pups. The effects of the sequence of the depression test (before or after the formalin test) on measures of depression were different in prenatally unstressed and stressed rat pups. In the former there were no differences between the two test sequences, while in prenatally stressed rat pups the first sequence showed a significant increase in the duration of immobility. The order of testing had no effect on the pain response--there were no differences between the numbers of flexion + shaking patterns in either prenatally stressed rat pups or unstressed animals; measures of the pain response were significantly greater in the sequence in which the formalin test was followed by the Porsolt test in prenatally stressed individuals as compared with unstressed animals. A positive correlation between study parameters was seen in the first series in prenatally unstressed rat pups, while there was a negative correlation in prenatally stressed animals. In the second series, there were no significant relationships between measures. Thus, the sequelae of postnatal stress, as imposed by each test the day before the final test, were apparent only in prenatally stressed animals in terms of the level of depression.
Collapse
|
26
|
Laprairie JL, Johns ME, Murphy AZ. Preemptive morphine analgesia attenuates the long-term consequences of neonatal inflammation in male and female rats. Pediatr Res 2008; 64:625-30. [PMID: 18679159 PMCID: PMC2638169 DOI: 10.1203/pdr.0b013e31818702d4] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Despite mounting evidence on the importance of pain management in preterm infants, clinical use of analgesics in this population is limited. Our previous studies have shown that neonatal inflammation results in long-term alterations in adult somatosensory thresholds, characterized by decreased baseline nociceptive sensitivity, and enhanced hyperalgesia after a subsequent inflammatory insult. The present studies were conducted to determine whether preemptive morphine attenuates these negative consequences. At P0, pups received an injection of morphine sulfate before an intraplantar injection of 1% carrageenan. Control pups received either saline (SAL) followed by intraplantar carrageenan, morphine sulfate followed by intraplantar SAL, or SAL followed by intraplantar SAL. Preemptive morphine significantly attenuated neonatal injury-induced hypoalgesia in adolescence and adulthood. Similarly, morphine pretreated animals displayed significantly less hyperalgesia and recovered faster from a subsequent inflammatory insult compared with controls. Neonatal morphine had no significant effect on morphine analgesia in adulthood. Interestingly, neonatally injured animals that did not receive morphine displayed a significant rightward shift in the morphine dose-response curve in the absence of peripheral inflammation. Together, these results demonstrate that preemptive morphine significantly attenuates the long-term behavioral impact of neonatal inflammatory injury.
Collapse
Affiliation(s)
- Jamie L Laprairie
- Department of Biology, Georgia State University, Atlanta, Georgia 30303-3088, USA
| | | | | |
Collapse
|
27
|
Bisogno T, Ortar G, Petrosino S, Morera E, Palazzo E, Nalli M, Maione S, Di Marzo V. Development of a potent inhibitor of 2-arachidonoylglycerol hydrolysis with antinociceptive activity in vivo. Biochim Biophys Acta Mol Cell Biol Lipids 2008; 1791:53-60. [PMID: 19027877 DOI: 10.1016/j.bbalip.2008.10.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 10/16/2008] [Accepted: 10/21/2008] [Indexed: 01/24/2023]
Abstract
Although inhibitors of the enzymatic hydrolysis of the endocannabinoid 2-arachidonoylglycerol are available, they are either rather weak in vitro (IC(50)>30 microM) or their selectivity towards other proteins of the endocannabinoid system has not been tested. Here we describe the synthesis and activity in vitro and in vivo of a tetrahydrolipstatin analogue, OMDM169, as a potent inhibitor of 2-AG hydrolysis, capable of enhancing 2-AG levels and of exerting analgesic activity via indirect activation of cannabinoid receptors. OMDM169 exhibited 0.13 microM<IC(50)<0.41 microM towards 2-AG hydrolysing activities in COS-7 cells and rat cerebellum, and inhibited (IC(50)=0.89 microM) the human recombinant MAGL, whilst being inactive (K(i)>10 microM) at human CB(1) and CB(2) receptors. However, OMDM169 shared with tetrahydrolipstatin the capability of inhibiting the human pancreatic lipase (IC(50)=0.6 microM). OMDM169 inhibited fatty acid amide hydrolase and diacylglycerol lipase only at higher concentrations (IC(50)=3.0 and 2.8 microM, respectively), and, accordingly, it increased by approximately 1.6-fold the levels of 2-AG, but not anandamide, in intact ionomycin-stimulated N18TG2 neuroblastoma cells. Acute intraperitoneal (i.p.) administration of OMDM169 to mice inhibited the second phase of the formalin-induced nocifensive response with an IC(50) of approximately 2.5 mg/kg, and concomitantly elevated 2-AG, but not anandamide, levels in the ipsilateral paw of formalin-treated mice. The antinociceptive effect of OMDM169 was antagonized by antagonists of CB(1) and CB(2) receptors, AM251 and AM630, respectively (1 mg/kg, i.p.). OMDM69 might represent a template for the development of selective and even more potent inhibitors of 2-AG hydrolysis.
Collapse
Affiliation(s)
- Tiziana Bisogno
- Institute of Biomolecular Chemistry, C.N.R., Pozzuoli (Naples), Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Himmel HM. Safety pharmacology assessment of central nervous system function in juvenile and adult rats: effects of pharmacological reference compounds. J Pharmacol Toxicol Methods 2008; 58:129-46. [PMID: 18585470 DOI: 10.1016/j.vascn.2008.06.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Accepted: 06/01/2008] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Recent EU/US pediatric legislation and FDA/EMEA guidelines recognize the potential differences in safety profiles of drugs in adults versus young patients. Hence safety studies are recommended to investigate key functional domains of e.g. the developing CNS. METHODS Selected psychoactive stimulants (caffeine, d-amphetamine, scopolamine) and depressants (baclofen, diazepam, haloperidol, chlorpromazine, imipramine, morphine) were characterized upon single administration with regard to behavioural parameters, locomotor activity, body temperature, pro-/anti-convulsive activity (pentylenetetrazole, PTZ), and nocifensive responses (hotplate) in neonatal (2 weeks), juvenile (4 weeks) and adult rats (8-9 weeks). RESULTS In vehicle-treated rats, behavioural patterns matured with age, locomotor activity and handling-induced rise in body temperature were enhanced, whereas PTZ convulsion threshold dose and nocifensive response latency decreased. Single test compound treatment elicited behavioural effects characteristic for psychoactive drugs with stimulating and depressing properties regardless of age. However, incidence of certain behaviours, and magnitude of effects on locomotor activity and body temperature varied with age and became generally more pronounced in adult rats. Pro-/anti-convulsive effects and delayed nocifensive responses did not differ between juvenile and adult rats. CONCLUSION CNS effects of selected psychoactive reference compounds were qualitatively similar, but quantitatively different in neonatal, juvenile and adult rats.
Collapse
Affiliation(s)
- Herbert M Himmel
- BHC-GDD-GED-NDS-SP, Safety Pharmacology, Bayer HealthCare AG, Wuppertal, Germany.
| |
Collapse
|
29
|
Anand KJS, Garg S, Rovnaghi CR, Narsinghani U, Bhutta AT, Hall RW. Ketamine reduces the cell death following inflammatory pain in newborn rat brain. Pediatr Res 2007; 62:283-90. [PMID: 17551412 DOI: 10.1203/pdr.0b013e3180986d2f] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Premature infants experience untreated repetitive pain that may alter their brain development. Effects of ketamine and repetitive pain on cellular death and subsequent behavior were studied in neonatal rats. Rat pups were randomized to undisturbed controls (C), 4% formalin injection (F), ketamine alone (K, 5 mg/kg) or formalin plus ketamine (KF) and were assessed for neuroactivation with Fos protein, cellular death with FluoroJade-B, cognition with the radial arm maze, and pain thresholds with the hot-plate. Greater Fos expression and cell death occurred in F vs. C groups in defined brain areas at 1 and 4 h in F compared with other groups. Cell death was accentuated 3.3-fold in cortical areas and 1.6-fold in subcortical areas in the F compared with the C group following repetitive pain and sacrifice 18-20 h later. These effects were ameliorated by ketamine. Compared with the F group, all other groups demonstrated greater exploratory and rearing behaviors and decreased time for bait consumption at 1-h and 3-h intervals. Significantly greater thermal pain latencies occurred in the KF and F groups. Repetitive neonatal pain accentuates neuronal excitation and cell death in developmentally regulated cortical and subcortical areas, which decreases the acquisition of visual-spatial clues, short-term and long-term memory, and increases pain latencies. Ketamine analgesia mitigates most of these effects.
Collapse
Affiliation(s)
- Kanwaljeet J S Anand
- Pain Neurobiology Lab, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas 72202, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Butkevich IP, Barr GA, Vershinina EA. Sex differences in formalin-induced pain in prenatally stressed infant rats. Eur J Pain 2007; 11:888-94. [PMID: 17379552 DOI: 10.1016/j.ejpain.2007.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2006] [Revised: 01/29/2007] [Accepted: 02/05/2007] [Indexed: 11/27/2022]
Abstract
The aim of this work was to study the effects of prenatal stress on nociceptive responses in the formalin test in female and male infant (7-day-old) Long-Evans hooded rats. Prenatally stressed infant rats displayed biphasic flinching+ shaking behavior whereas non-stressed animals showed only a weak second phase. Pain sensitivity in prenatally stressed males was significantly greater than that of prenatally non-stressed males during the second phase only; there were no differences in pain sensitivity between prenatally stressed and non-stressed females. Moreover prenatally stressed male rats pups demonstrated that the second phase of the response to formalin was enhanced relative to the second phase in stressed females. The current and previous data [Butkevich IP, Barr GA, Mikhailenko VA, Otellin VA. Increased formalin-induced pain and expression of fos neurons in the lumbar spinal cord of prenatally stressed infants rats. Neurosci Lett 2006a;403:222-226] show increased tonic pain in prenatally stressed infant rats and a large increase in the number of formalin-induced fos-like immunoreactivity in the spinal cord dorsal horn. There is a concomitant decrease in serotonin-like immunoreactivity in the lumbar spinal cord dorsal horn [Butkevich IP, Barr GA, Otellin VA. Effect of prenatal stress on behavioral and neural indices of formalin-induced pain in infant rats. Abstracts, 35th Annual Meeting of Soc. For Neurosci. 2005a. Program No. 512.4 Washington, DC: Society for Neuroscience]. Given the decreased level of perinatal testosterone in prenatally stressed rats to which infant males are more sensitive than females, we suggest that these hormonal, behavioral and neuronal indices are strongly interrelated in prenatally stressed 7-day-old rat pups and that the decreased surge of testosterone may contribute to the increased behavioral response in the second phase in male rat pups. Mechanisms underlying the behavioral pain response induced by inflammation in prenatally stressed rat pups are characterized by sexual dimorphism even prior to the activational effects of sex hormones.
Collapse
Affiliation(s)
- Irina P Butkevich
- Laboratory of Ontogeny of Nervous System, I.P. Pavlov Institute of Physiology, The Russian Academy of Sciences, St. Petersburg 199034, Russia.
| | | | | |
Collapse
|
31
|
Koepp J, Lindsey CJ, Motta EM, Rae GA. Role of the paratrigeminal nucleus in nocifensive responses of rats to chemical, thermal and mechanical stimuli applied to the hind paw. Pain 2006; 122:235-244. [PMID: 16616416 DOI: 10.1016/j.pain.2006.01.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2005] [Revised: 12/16/2005] [Accepted: 01/25/2006] [Indexed: 11/29/2022]
Abstract
Anatomical and immunohystochemical data suggest the paratrigeminal nucleus (Pa5) may play a role in nociceptive processing. The current study examines the influence of unilateral Pa5 lesion on nocifensive responses of conscious rats to noxious thermal (Hargreaves test), mechanical (electronic von Frey and Randall-Selitto tests), and chemical (formalin 2.5%; 50 microl) stimuli applied to the hind paw. Lesion of the Pa5 induced by ibotenic acid did not affect the latency for radiant heat-induced withdrawal of either paw. In contrast, the mean mechanical threshold for withdrawal of the contralateral (but not ipsilateral) paw in Pa5-lesioned rats was reduced by approximately 45% and 20%, in electronic von Frey and Randall-Selitto tests, respectively, when compared to sham-operated animals. Conversely, animals with Pa5 lesions injected with formalin in the contralateral paw spent less time engaged in focused (licking, biting or scratching the injected paw) and total nocifensive behavior (i.e., focused nocifensive behavior plus protection of the injected paw during movements) in both the first and second phases of the test [ approximately 50% inhibition of each parameter during first phase (0-5 min) and at 20, 25, and 30 min of second phase, relative to the sham-operated group], but the number of paw-jerks was unaffected. Pa5 lesion also delayed the onset of second phase focused pain induced by formalin in the ipsilateral paw. The results suggest that the Pa5 integrates the supraspinal pain control system and plays a differential modulatory role in the central processing of mechanical and chemical nociceptive information.
Collapse
Affiliation(s)
- Janice Koepp
- Department of Pharmacology, Biological Sciences Center, Universidade Federal de Santa Catarina, Campus, Trindade, Florianópolis, SC 88010-970, Brazil Department of Biophysics, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil
| | | | | | | |
Collapse
|
32
|
Zissen MH, Zhang G, Kendig JJ, Sweitzer SM. Acute and chronic morphine alters formalin pain in neonatal rats. Neurosci Lett 2006; 400:154-7. [PMID: 16517072 DOI: 10.1016/j.neulet.2006.02.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2005] [Revised: 02/14/2006] [Accepted: 02/14/2006] [Indexed: 11/30/2022]
Abstract
The present study tested the hypothesis that morphine exposure during the human developmental equivalent of the third trimester would alter inflammatory pain. This study examined whether acute or continuous opioid exposure in the neonatal rat alters formalin-induced nociception after 4 days of abstinence. Rats were exposed to a single acute administration of morphine on postnatal day 7 or 72 h of opioid infusion from postnatal days 5-7 via osmotic pump. When challenged with intraplantar formalin on postnatal day 11, rats exposed to acute or chronic morphine had increased phase II pain-associated behaviors. These findings suggest that neonatal morphine exposure may have unintended consequences on inflammatory pain.
Collapse
Affiliation(s)
- Maurice H Zissen
- Department of Anesthesiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
33
|
Abstract
The study of pain development has come into its own. Reaping the rewards of years of developmental and molecular biology, it has now become possible to translate fundamental knowledge of signalling pathways and synaptic physiology into a better understanding of infant pain. Research has cast new light on the physiological and pharmacological processes that shape the newborn pain response, which will help us to understand early pain behaviour and to design better treatments. Furthermore, it has shown how developing pain circuitry depends on non-noxious sensory activity in the healthy newborn, and how early injury can permanently alter pain processing.
Collapse
Affiliation(s)
- Maria Fitzgerald
- Department of Anatomy and Developmental Biology, Wellcome Pain Consortium; University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
34
|
Carbajal R, Lenclen R, Jugie M, Paupe A, Barton BA, Anand KJS. Morphine does not provide adequate analgesia for acute procedural pain among preterm neonates. Pediatrics 2005; 115:1494-500. [PMID: 15930209 DOI: 10.1542/peds.2004-1425] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Morphine alleviates prolonged pain, reduces behavioral and hormonal stress responses induced by surgery among term neonates, and improves ventilator synchrony and sedation among ventilated preterm neonates, but its analgesic effects on the acute pain caused by invasive procedures remain unclear. OBJECTIVE To investigate the analgesic efficacy of intravenously administered morphine on heel stick-induced acute pain among preterm neonates. DESIGN This study was nested within a prospective, randomized, double-blind, multicenter, placebo-controlled trial (the NEOPAIN Trial). SETTING A tertiary-care NICU in a teaching hospital. PARTICIPANTS Forty-two preterm neonates undergoing ventilation. INTERVENTIONS Neonates were randomized to either the morphine (loading dose of 100 microg/kg, followed by infusions of 10-30 microg/kg per hour according to gestation, N = 21) or placebo (5% dextrose infusions, N = 21) group. Pain responses to 3 heel sticks were evaluated, ie, before the loading dose (T1), 2 to 3 hours after the loading dose (T2), and 20 to 28 hours after the loading dose (T3). MAIN OUTCOMES MEASURES Pain was assessed with the Douleur Aiguë Nouveau-né (DAN) scale (behavioral pain scale) and the Premature Infant Pain Profile (PIPP) (multidimensional pain scale); plasma morphine levels were measured at T3. RESULTS Infants in the placebo and morphine groups had similar gestational ages (mean +/- SD: 27.2 +/- 1.7 vs 27.3 +/- 1.8 weeks) and birth weights (972 +/- 270 vs 947 +/- 269 g). Mean +/- SD DAN pain scores at T1, T2, and T3 were 4.8 +/- 4.0, 4.6 +/- 2.9, and 4.7 +/- 3.6, respectively, for the placebo group and 4.5 +/- 3.8, 4.4 +/- 3.7, and 3.1 +/- 3.4 for the morphine group. The within-group factor (pain at T1, T2, and T3) was not statistically different over time. The between-group analysis (infants receiving placebo versus those receiving morphine) showed no significant differences. Mean +/- SD PIPP pain scores at T1, T2, and T3 were 11.5 +/- 4.8, 11.1 +/- 3.7, and 9.1 +/- 4.0, respectively, for the placebo group and 10.0 +/- 3.6, 8.8 +/- 4.9, and 7.8 +/- 3.6 for the morphine group. The within-group factor was statistically different over time. The between-group analysis showed no significant differences. Mean +/- SD plasma morphine levels at T3 were 0.44 +/- 1.79 ng/mL and 63.36 +/- 33.35 ng/mL for the placebo and morphine groups, respectively. There was no correlation between plasma morphine levels and pain scores at T3 (DAN, R = -0.05; PIPP, R = -0.02). CONCLUSIONS Despite its routine use in the NICU, morphine given as a loading dose followed by continuous intravenous infusions does not appear to provide adequate analgesia for the acute pain caused by invasive procedures among ventilated preterm neonates.
Collapse
MESH Headings
- Acute Disease
- Analgesics, Opioid/blood
- Analgesics, Opioid/therapeutic use
- Blood Specimen Collection/adverse effects
- Double-Blind Method
- Female
- Gestational Age
- Humans
- Hypnotics and Sedatives/blood
- Hypnotics and Sedatives/therapeutic use
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/blood
- Infant, Premature, Diseases/drug therapy
- Infusions, Intravenous
- Male
- Morphine/blood
- Morphine/therapeutic use
- Pain/drug therapy
- Pain/etiology
- Pain Measurement
- Prospective Studies
- Respiration, Artificial
Collapse
Affiliation(s)
- Ricardo Carbajal
- Neonatal Intensive Care Unit, Poissy Saint Germain Hospital, Poissy, France.
| | | | | | | | | | | |
Collapse
|
35
|
Taneja I, Bruehl S, Robertson D. Effect of modafinil on acute pain: a randomized double-blind crossover study. J Clin Pharmacol 2005; 44:1425-7. [PMID: 15545315 DOI: 10.1177/0091270004270292] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Indu Taneja
- Autonomic Dysfunction Center, Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University, Nashville, TN 37232-2195, USA
| | | | | |
Collapse
|
36
|
Teixeira CFP, Landucci ECT, Antunes E, Chacur M, Cury Y. Inflammatory effects of snake venom myotoxic phospholipases A2. Toxicon 2004; 42:947-62. [PMID: 15019493 DOI: 10.1016/j.toxicon.2003.11.006] [Citation(s) in RCA: 164] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Snake venom phospholipases A2 (PLA2) show a remarkable functional diversity. Among their toxic activities, some display the ability to cause rapid necrosis of skeletal muscle fibers, thus being myotoxic PLA2s. Besides myotoxicity, these enzymes evoke conspicuous inflammatory and nociceptive events in experimental models. Local inflammation and pain are important characteristics of snakebite envenomations inflicted by viperid and crotalid species, whose venoms are rich sources of myotoxic PLA2s. Since the discovery that mammalian PLA2 is a key enzyme in the release of arachidonic acid, the substrate for the synthesis of several lipid inflammatory mediators, much interest has been focused on this enzyme in the context of inflammation. The mechanisms involved in the proinflammatory action of secretory PLA2s are being actively investigated, and part of the knowledge on secretory PLA2 effects has been gained by using snake venom PLA2s as tools, due to their high structural homology with human secretory PLA2s. The inflammatory events evoked by PLA2s are primarily associated with enzymatic activity and to the release of arachidonic acid metabolites. However, catalytically inactive Lys49 PLA2s trigger inflammatory and nociceptive responses comparable to those of their catalytically active counterparts, thereby evidencing that these proteins promote inflammation and pain by mechanisms not related to phospholipid hydrolysis nor to mobilization of arachidonic acid. These studies have provided a boost to the research in this field and various approaches have been used to identify the amino acid residues and the specific sites of interaction of myotoxic PLA2s with cell membranes potentially involved in the PLA2-induced inflammatory and nociceptive effects. This work reviews the proinflammatory and nociceptive effects evoked by myotoxic PLA2s and their mechanisms of action.
Collapse
Affiliation(s)
- C F P Teixeira
- Laboratories of Pharmacology, Instituto Butantan, 05503-900 São Paulo, SP, Brazil.
| | | | | | | | | |
Collapse
|
37
|
Hakki Onen S, Alloui A, Jourdan D, Eschalier A, Dubray C. Effects of rapid eye movement (REM) sleep deprivation on pain sensitivity in the rat. Brain Res 2001; 900:261-7. [PMID: 11334806 DOI: 10.1016/s0006-8993(01)02320-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The relationship between pain and sleep seems to be reciprocal: if pain may interrupt or disturb sleep, poor sleep can also influence pain perception. However the influence of sleep disturbances on pain sensitivity remain poorly investigated. The aim of this study was to assess the effect of REM sleep deprivation on the reaction of rats subjected to different noxious stimuli. In each experiment 16 Wistar male rats were randomly assigned to two groups: controls (n=8), and REM sleep deprived rats (n=8). REM sleep deprivation was elicited using the 'inverted flower pot' technique. Four different experiments were performed to assess the sensitivity to mechanical (vocalization threshold in paw pressure), thermal (tail withdrawal latency in hot water immersion), electrical (envelope of 2nd peep in tail shock test) and chemical (analgesic behavior in formalin test) noxious stimuli. All experiments were performed over a 5-day period with baseline (day 1, day 2) in a dry environment and REM sleep deprivation (day 3, day 4 and day 5) in a wet environment. Under wet conditions, vocalization threshold in the paw pressure test (-20%, P=0.005), and tail withdrawal latency in the hot water immersion test (-21%, P=0.006) were significantly lower, and the envelope of 2nd peep in the tail electrical shock was significantly greater (+78%, P=0.009), in REM sleep deprived rats compared to controls. However, under wet conditions the mean duration of nociceptive behaviors in the formalin test did not differ between the two groups. In conclusion, REM sleep deprivation induces a significant increase in the behavioral responses to noxious mechanical, thermal and electrical stimuli in rats.
Collapse
Affiliation(s)
- S Hakki Onen
- Laboratoire de Pharmacologie Médicale, INSERM, EMI-HU 9904, Faculté de Médecine B.P. 38, 63001 Cedex 1, Clermont-Ferrand, France.
| | | | | | | | | |
Collapse
|
38
|
Abstract
We investigated the scoring properties of the mouse formalin test using the time-sampling method recently developed for infant and adult rats. Formalin was injected under the plantar surface of one rear paw (10 microl, 1-8%), and pain behaviours (paw favouring, lifting and licking) and behavioural state were recorded. Correlational and regression analyses indicated that scores composed of combinations of all three pain behaviours, either summed or weighted, provided less variable indices of pain than licking alone. The maximum percent effect (MPE(50); i.e. pain behaviour 50% of the time) for the log formalin concentration-effect curves was 3-4% in both phases. Habituation to the test environment prior to testing did not alter the MPE(50)s, but slopes were lower in unhabituated mice, dramatically increasing the size of the confidence interval. Formalin dose-dependently reduced locomotion, rearing and sniffing in both the first phase and the early part of the second phase. The combination measures were sensitive to morphine (2-8 mg/kg), amphetamine (1-4 mg/kg), dipyrone (50-200 mg/kg), xylazine (0.25-1 mg/kg), and acepromazine (0.25-1 mg/kg), and resistant to diazepam (0.5-2 mg/kg), pimozide (0.05-0.25 mg/kg), pentobarbital (10 and 15 mg/kg) and indomethacin (2-8 mg/kg). Decreased pain was correlated with increased motor activity for morphine and amphetamine, and with decreased activity for xylazine and acepromazine; dipyrone and indomethacin did not alter activity levels.
Collapse
Affiliation(s)
- Ghada-Maria Saddi
- Department of Family Medicine, McGill University, 515-517 Pine Avenue West, Montreal, QC, H2W 1S4, Canada Department of Psychiatry, McGill University, 1033 Pine Avenue West, Montreal, QC, H3A 1A1, Canada
| | | |
Collapse
|
39
|
|
40
|
|
41
|
Abstract
A time-sampling method that allows up to eight rats to be tested simultaneously in the formalin test is described and compared to the continuous rating method. Time sampling the behavioural response to formalin every 1 or 2 min produces scores that are essentially identical to continuous rating for both the formalin concentration effect relationship and the morphine dose effect relationship, with no loss of statistical power. The most important advantage of the method is that it allows data on other aspects of the rats' behaviour, such as behavioural state and the side effects of drugs to be scored during the formalin test. Formalin injection produces a dose-dependent decrease in locomotor and exploratory activity. The activity pattern of rats is normalized at morphine doses that produce about a 50% reduction in pain, while morphine doses high enough to completely suppress the pain response are accompanied by considerable sedation. The use of the jackknifing procedure to obtain unbiased estimates of the variability of parameters estimated from dose effect relationships is also described.
Collapse
Affiliation(s)
- Frances V Abbott
- Department of Psychiatry, McGill University, 1033 Pine Ave West, Montreal, QC H3A 1A1, Canada Department of Psychology, McGill University, 1205 Dr. Penfield Ave, Montreal, QC H3A 1B1, Canada
| | | | | | | |
Collapse
|
42
|
Abstract
We report the results of a study designed to assess age differences in the response to the formalin test, a model of tissue injury and inflammation, while controlling for differences in weight and motoric abilities in three groups of adult male Long-Evans rats: young (3 months old), middle-aged (18 months old), and old (24 months old). The first part of the study assessed initial differences in responsivity and found that the middle-aged group showed the greatest response, whereas the young and old groups did not differ from each other. In the second part of the study, the young and middle-aged animals were followed for a 4-month period. The formalin test was repeated at 2-month intervals. These results indicate that there may be an age-associated change in the sensitivity to tonic pain and that this sensitivity may peak at mid-life.
Collapse
Affiliation(s)
- L Gagliese
- Department of Psychology, McGill University, Montreal, Quebec, Canada.
| | | |
Collapse
|
43
|
Rahman W, Dickenson AH. Electrophysiological studies on the postnatal development of the spinal antinociceptive effects of the delta opioid receptor agonist DPDPE in the rat. Br J Pharmacol 1999; 126:1115-22. [PMID: 10204998 PMCID: PMC1565889 DOI: 10.1038/sj.bjp.0702418] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
1. The antinociceptive effects of the delta opioid receptor selective agonist, DPDPE [(D-Pen2,D-Pen5)-enkephalin] was studied in rats aged postnatal day (P) 14, P21, P28 and P56. 2. Antinociceptive effects of DPDPE were measured as percentage inhibition of the C-fibre evoked response and post-discharge of dorsal horn neurones evoked by peripheral electrical stimulation. DPDPE was administered by topical application, akin to intrathecal injection. 3. DPDPE (0.1-100 microg) produced dose-related inhibitions at all ages; these inhibitions were reversed by 5 microg of the opioid antagonist naloxone. 4. The dose-response curves for C-fibre evoked response and post-discharge of the neurones were not different in rats aged P14 and P21. DPDPE was significantly more potent at P14 and P21 compared with its inhibitory effects on these responses at P28 and P56. 5. DPDPE produced minor inhibitions of the A-fibre evoked response of the neurones at P14, P21, P28 and P56, suggesting that the inhibitory effects of DPDPE are mediated via presynaptic receptors on the terminals of C-fibre afferents. 6. Since spinal delta opioid receptor density changes little over this period, the increased antinociceptive potency of DPDPE in the rat pups compared with the adult is likely to be due to post-receptor events, or in developmental changes in the actions of other transmitter/receptor systems within the spinal cord.
Collapse
Affiliation(s)
- W Rahman
- Department of Pharmacology, University College London, England
| | | |
Collapse
|
44
|
Rahman W, Dashwood MR, Fitzgerald M, Aynsley-Green A, Dickenson AH. Postnatal development of multiple opioid receptors in the spinal cord and development of spinal morphine analgesia. BRAIN RESEARCH. DEVELOPMENTAL BRAIN RESEARCH 1998; 108:239-54. [PMID: 9693800 DOI: 10.1016/s0165-3806(98)00054-6] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The postnatal ontogeny of mu, delta and kappa opioid receptor binding sites in the spinal cord of rat pups at various postnatal days was determined using in vitro autoradiographical methods. The functional effect of spinal morphine was also assessed using in vivo electrophysiological methods in rats at P14, P21 and adults (P56). Both mu and kappa opioid receptor binding-sites are present from P0 and spread relatively diffusely throughout the spinal cord. Overall binding peaks at P7 and subsequently decreases to adult levels with the mu opioid receptor binding sites regressing to become denser in the superficial dorsal horn. delta Opioid receptor binding was first seen at P7, and no distinction between superficial and deeper laminae was seen. In the adult, the relative proportions of the opiate receptors in the superficial dorsal horn are 63%, 22% and 15%, for mu, delta and kappa receptor binding sites, respectively. C-fibre evoked dorsal horn neuronal responses recorded from anaesthetized rat pups were highly sensitive to spinal morphine at P21, (EC50 0.005 microgram), compared to the adult (EC50 0.9 microgram). However, the EC50 (0.2 microgram) at P14 was greater than at P21 despite the fact that mu receptor binding was greater at P14. Opioid receptor binding is developmentally regulated and undergoes substantial postnatal reorganization. However, the number of mu receptor binding sites appears not to be the only determinant of functional sensitivity to spinal morphine. Other factors, such as coupling of the receptors are likely to be important.
Collapse
MESH Headings
- Analgesics/pharmacology
- Analgesics, Opioid/pharmacology
- Animals
- Autoradiography
- Benzeneacetamides
- Dose-Response Relationship, Drug
- Electrophysiology
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-
- Enkephalin, D-Penicillamine (2,5)-
- Enkephalins/pharmacology
- Male
- Morphine/pharmacology
- Nerve Fibers/chemistry
- Nerve Fibers/drug effects
- Nerve Fibers/physiology
- Pyrrolidines/pharmacology
- Radioligand Assay
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid/agonists
- Receptors, Opioid/analysis
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/analysis
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/analysis
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/analysis
- Spinal Cord/chemistry
- Spinal Cord/drug effects
- Spinal Cord/growth & development
Collapse
Affiliation(s)
- W Rahman
- Dept. of Pharmacology, University College London, UK
| | | | | | | | | |
Collapse
|
45
|
Abstract
We investigated the behavioral response of rat pups to intraplantar injection of varying formalin concentrations using a time-sampling method. At 3 days of age, the response was monophasic and persisted for the whole hour, even at low formalin concentrations. Flexion, shaking and licking the injected limb and hind-limb kicking correlated strongly with log formalin concentration (r = 0.82); behavioral state was altered only at the highest concentration. The response on day 15 was also monophasic, but it waned in 30 min, even at the highest formalin concentration tested. Flexion, shaking and licking of the injected limb were strong pain measures (r = 0.83). The response at 25 days was biphasic, and the adult measures, paw lifting and licking, produced a good formalin concentration-effect relationship (r = 0.80). The log concentration-effect relationships for formalin at the three developmental stages and for adult rats were parallel, but between 3 days and 15 days of age, the relationship shifted to the right by 2.5-fold, and by a further 4-fold between 15 and 25 days, when the sensitivity to formalin-induced pain was similar to that in adults. The data describe efficient, quantitative measures of formalin-induced pain for developing rats, show that the pain response is log-linearly related to formalin concentration throughout development, and demonstrate that the sensitivity to formalin-induced pain is about 10-fold higher in neonatal rats than in weanlings. the data imply that there are major qualitative changes in pain processing as the nervous system develops.
Collapse
Affiliation(s)
- Carolyn J Teng
- Department of Psychiatry, McGill University, 1033 Pine Ave. West, Montreal, QC H3A 1A1, Canada
| | | |
Collapse
|
46
|
|
47
|
Affiliation(s)
- K Andrews
- Department of Anatomy and Developmental Biology, University College London, England
| | | |
Collapse
|
48
|
Abstract
This article is the eighteenth installment of our annual review of research concerning the opiate system. It includes articles published during 1995 reporting the behavioral effects of the opiate peptides and antagonists, excluding the purely analgesic effects. The specific topics covered this year include stress: tolerance and dependence; eating; drinking; gastrointestinal, renal, and hepatic function; mental illness and mood; learning, memory, and reward; cardiovascular responses; respiration and thermoregulation; seizures and other neurological disorders; electrical-related activity; general activity and locomotion; sex, pregnancy, and development; immunological responses; and other behaviors.
Collapse
Affiliation(s)
- G A Olson
- Department of Psychology, University of New Orleans, LA 70148, USA
| | | | | |
Collapse
|